Long-Lived Antitumor CD8+ Lymphocytes for Adoptive Therapy Generated Using an Artificial Antigen-Presenting Cell

MO Butler, JS Lee, S Ansen, D Neuberg, FS Hodi… - Clinical Cancer …, 2007 - AACR
MO Butler, JS Lee, S Ansen, D Neuberg, FS Hodi, AP Murray, L Drury, A Berezovskaya…
Clinical Cancer Research, 2007AACR
Purpose: Antitumor lymphocytes can be generated ex vivo unencumbered by
immunoregulation found in vivo. Adoptive transfer of these cells is a promising therapeutic
modality that could establish long-term antitumor immunity. However, the widespread use of
adoptive therapy has been hampered by the difficulty of consistently generating potent
antitumor lymphocytes in a timely manner for every patient. To overcome this, we sought to
establish a clinical grade culture system that can reproducibly generate antigen-specific …
Abstract
Purpose: Antitumor lymphocytes can be generated ex vivo unencumbered by immunoregulation found in vivo. Adoptive transfer of these cells is a promising therapeutic modality that could establish long-term antitumor immunity. However, the widespread use of adoptive therapy has been hampered by the difficulty of consistently generating potent antitumor lymphocytes in a timely manner for every patient. To overcome this, we sought to establish a clinical grade culture system that can reproducibly generate antigen-specific cytotoxic T lymphocytes (CTL).
Experimental Design: We created an off-the-shelf, standardized, and renewable artificial antigen-presenting cell (aAPC) line that coexpresses HLA class I, CD54, CD58, CD80, and the dendritic cell maturation marker CD83. We tested the ability of aAPC to generate tumor antigen-specific CTL under optimal culture conditions. The number, phenotype, effector function, and in vitro longevity of generated CTL were determined.
Results: Stimulation of CD8+ T cells with peptide-pulsed aAPC generated large numbers of functional CTL that recognized a variety of tumor antigens. These CTLs, which possess a phenotype consistent with in vivo persistence, survived ex vivo for prolonged periods of time. Clinical grade aAPC33, produced under current Good Manufacturing Practices guidelines, generated sufficient numbers of CTL within a short period of time. These CTL specifically lysed a variety of melanoma tumor lines naturally expressing a target melanoma antigen. Furthermore, antitumor CTL were easily generated in all melanoma patients examined.
Conclusions: With clinical grade aAPC33 in hand, we are now poised for clinical translation of ex vivo generated antitumor CTL for adoptive cell transfer.
AACR